Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 43(18): 8990-9005, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26261209

ABSTRACT

In recent times, high-throughput screening analyses have broadly defined the RNA cellular targets of TDP-43, a nuclear factor involved in neurodegeneration. A common outcome of all these studies is that changing the expression levels of this protein can alter the expression of several hundred RNAs within cells. What still remains to be clarified is which changes represent direct cellular targets of TDP-43 or just secondary variations due to the general role played by this protein in RNA metabolism. Using an HTS-based splicing junction analysis we identified at least six bona fide splicing events that are consistent with being controlled by TDP-43. Validation of the data, both in neuronal and non-neuronal cell lines demonstrated that TDP-43 substantially alters the levels of isoform expression in four genes potentially important for neuropathology: MADD/IG20, STAG2, FNIP1 and BRD8. For MADD/IG20 and STAG2, these changes could also be confirmed at the protein level. These alterations were also observed in a cellular model that successfully mimics TDP-43 loss of function effects following its aggregation. Most importantly, our study demonstrates that cell cycle alterations induced by TDP-43 knockdown can be recovered by restoring the STAG2, an important component of the cohesin complex, normal splicing profile.


Subject(s)
Alternative Splicing , Antigens, Nuclear/genetics , DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Antigens, Nuclear/metabolism , Apoptosis , Binding Sites , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , DNA-Binding Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Mitosis , Protein Isoforms/genetics
2.
PLoS One ; 8(9): e74216, 2013.
Article in English | MEDLINE | ID: mdl-24098639

ABSTRACT

Current B-cell disorder treatments take advantage of dose-intensive chemotherapy regimens and immunotherapy via use of monoclonal antibodies. Unfortunately, they may lead to insufficient tumor distribution of therapeutic agents, and often cause adverse effects on patients. In this contribution, we propose a novel therapeutic approach in which relatively high doses of Hydroxychloroquine and Chlorambucil were loaded into biodegradable nanoparticles coated with an anti-CD20 antibody. We demonstrate their ability to effectively target and internalize in tumor B-cells. Moreover, these nanoparticles were able to kill not only p53 mutated/deleted lymphoma cell lines expressing a low amount of CD20, but also circulating primary cells purified from chronic lymphocitic leukemia patients. Their safety was demonstrated in healthy mice, and their therapeutic effects in a new model of Burkitt's lymphoma. The latter serves as a prototype of an aggressive lympho-proliferative disease. In vitro and in vivo data showed the ability of anti-CD20 nanoparticles loaded with Hydroxychloroquine and Chlorambucil to increase tumor cell killing in comparison to free cytotoxic agents or Rituximab. These results shed light on the potential of anti-CD20 nanoparticles carrying Hydroxychloroquine and Chlorambucil for controlling a disseminated model of aggressive lymphoma, and lend credence to the idea of adopting this therapeutic approach for the treatment of B-cell disorders.


Subject(s)
Antigens, CD20/therapeutic use , Chlorambucil/pharmacology , Disease Models, Animal , Hydroxychloroquine/pharmacology , Lymphoma, B-Cell/drug therapy , Nanoparticles/therapeutic use , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/immunology , Apoptosis/drug effects , Autophagy/drug effects , Cell Survival/drug effects , Chlorambucil/therapeutic use , Drug Combinations , Drug Delivery Systems/methods , Female , Flow Cytometry , Hydroxychloroquine/therapeutic use , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Electron, Transmission , Rituximab
3.
Nat Struct Mol Biol ; 20(3): 347-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396353

ABSTRACT

Topoisomerase I (TOP1) inhibitors are an important class of anticancer drugs. The cytotoxicity of TOP1 inhibitors can be modulated by replication fork reversal through a process that requires poly(ADP-ribose) polymerase (PARP) activity. Whether regressed forks can efficiently restart and what factors are required to restart fork progression after fork reversal are still unknown. We have combined biochemical and EM approaches with single-molecule DNA fiber analysis to identify a key role for human RECQ1 helicase in replication fork restart after TOP1 inhibition that is not shared by other human RecQ proteins. We show that the poly(ADP-ribosyl)ation activity of PARP1 stabilizes forks in the regressed state by limiting their restart by RECQ1. These studies provide new mechanistic insights into the roles of RECQ1 and PARP in DNA replication and offer molecular perspectives to potentiate chemotherapeutic regimens based on TOP1 inhibition.


Subject(s)
DNA Replication , RecQ Helicases/metabolism , Topoisomerase I Inhibitors/pharmacology , Camptothecin/pharmacology , Cell Line , DNA Topoisomerases, Type I/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RecQ Helicases/genetics
4.
Genome Res ; 23(1): 1-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23187890

ABSTRACT

We report the genome-wide mapping of ORC1 binding sites in mammals, by chromatin immunoprecipitation and parallel sequencing (ChIP-seq). ORC1 binding sites in HeLa cells were validated as active DNA replication origins (ORIs) using Repli-seq, a method that allows identification of ORI-containing regions by parallel sequencing of temporally ordered replicating DNA. ORC1 sites were universally associated with transcription start sites (TSSs) of coding or noncoding RNAs (ncRNAs). Transcription levels at the ORC1 sites directly correlated with replication timing, suggesting the existence of two classes of ORIs: those associated with moderate/high transcription levels (≥1 RNA copy/cell), firing in early S and mapping to the TSSs of coding RNAs; and those associated with low transcription levels (<1 RNA copy/cell), firing throughout the entire S and mapping to TSSs of ncRNAs. These findings are compatible with a scenario whereby TSS expression levels influence the efficiency of ORC1 recruitment at G(1) and the probability of firing during S.


Subject(s)
DNA Replication Timing , Genome, Human , Origin Recognition Complex/metabolism , Replication Origin/genetics , Transcription, Genetic , CD4-Positive T-Lymphocytes , Chromatin Immunoprecipitation , G1 Phase/genetics , Gene Expression Regulation , HeLa Cells , Humans , Origin Recognition Complex/genetics , Physical Chromosome Mapping , RNA, Untranslated/metabolism , S Phase/genetics , Transcription Initiation Site
5.
Mol Cancer ; 10: 83, 2011 Jul 13.
Article in English | MEDLINE | ID: mdl-21752281

ABSTRACT

BACKGROUND: RecQ helicases play an essential role in the maintenance of genome stability. In humans, loss of RecQ helicase function is linked with predisposition to cancer and/or premature ageing. Current data show that the specific depletion of the human RECQ1 helicase leads to mitotic catastrophe in cancer cells and inhibition of tumor growth in mice. RESULTS: Here, we show that RECQ1 is highly expressed in various types of solid tumors. However, only in the case of brain gliomas, the high expression of RECQ1 in glioblastoma tissues is paralleled by a lower expression in the control samples due to the poor expression of RECQ1 in non-dividing tissues. This conclusion is validated by immunohistochemical analysis of a tissue microarray containing 63 primary glioblastomas and 19 perilesional tissue samples, as control. We also show that acute depletion of RECQ1 by RNAi results in a significant reduction of cellular proliferation, perturbation of S-phase progression, and spontaneous γ-H2AX foci formation in T98G and U-87 glioblastoma cells. Moreover, RECQ1 depleted T98G and U-87 cells are hypersensitive to HU or temozolomide treatment. CONCLUSIONS: Collectively, these results indicate that RECQ1 has a unique and important role in the maintenance of genome integrity. Our results also suggest that RECQ1 might represent a new suitable target for anti cancer therapies aimed to arrest cell proliferation in brain gliomas.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation , Glioblastoma/genetics , RecQ Helicases/genetics , RecQ Helicases/physiology , Tumor Burden/genetics , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Middle Aged , RNA, Small Interfering/pharmacology , RecQ Helicases/antagonists & inhibitors , RecQ Helicases/metabolism , Tumor Burden/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
6.
Nucleic Acids Res ; 39(5): 1703-17, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21059676

ABSTRACT

RecQ helicases have attracted considerable interest in recent years due to their role in the suppression of genome instability and human diseases. These atypical helicases exert their function by resolving a number of highly specific DNA structures. The crystal structure of a truncated catalytic core of the human RECQ1 helicase (RECQ1(49-616)) shows a prominent ß-hairpin, with an aromatic residue (Y564) at the tip, located in the C-terminal winged-helix domain. Here, we show that the ß-hairpin is required for the DNA unwinding and Holliday junction (HJ) resolution activity of full-length RECQ1, confirming that it represents an important determinant for the distinct substrate specificity of the five human RecQ helicases. In addition, we found that the ß-hairpin is required for dimer formation in RECQ1(49-616) and tetramer formation in full-length RECQ1. We confirmed the presence of stable RECQ1(49-616) dimers in solution and demonstrated that dimer formation favours DNA unwinding; even though RECQ1 monomers are still active. Tetramers are instead necessary for more specialized activities such as HJ resolution and strand annealing. Interestingly, two independent protein-protein contacts are required for tetramer formation, one involves the ß-hairpin and the other the N-terminus of RECQ1, suggesting a non-hierarchical mechanism of tetramer assembly.


Subject(s)
DNA/metabolism , RecQ Helicases/chemistry , DNA, Cruciform , Dimerization , Humans , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , RecQ Helicases/metabolism
7.
PLoS One ; 5(11): e13720, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-21085491

ABSTRACT

BACKGROUND: The retinoblastoma protein (Rb) is a crucial regulator of cell cycle progression by binding with E2F transcription factor and repressing the expression of a variety of genes required for the G1-S phase transition. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that Rb and E2F1 directly participate in the control of initiation of DNA replication in human HeLa, U2OS and T98G cells by specifically binding to origins of DNA replication in a cell cycle regulated manner. We show that, both in vitro and inside the cells, the largest subunit of the origin recognition complex (Orc1) specifically binds hypo-phosphorylated Rb and that this interaction is competitive with the binding of Rb to E2F1. The displacement of Rb-bound Orc1 by E2F1 at origins of DNA replication marks the progression of the G1 phase of the cell cycle toward the G1-S border. CONCLUSIONS/SIGNIFICANCE: The participation of Rb and E2F1 in the formation of the multiprotein complex that binds origins of DNA replication in mammalian cells appears to represent an effective mechanism to couple the expression of genes required for cell cycle progression to the activation of DNA replication.


Subject(s)
DNA Replication/genetics , Origin Recognition Complex/metabolism , Replication Origin/genetics , Retinoblastoma Protein/metabolism , Binding, Competitive , Cell Line, Tumor , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Fluorescence Resonance Energy Transfer , G1 Phase/genetics , HeLa Cells , Humans , Immunoblotting , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Origin Recognition Complex/genetics , Phosphorylation , Protein Binding , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/genetics
8.
Nucleic Acids Res ; 38(22): 8105-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20693533

ABSTRACT

Recent evidence points to homeotic proteins as actors in the crosstalk between development and DNA replication. The present work demonstrates that HOXC13, previously identified as a new member of human DNA replicative complexes, is a stable component of early replicating chromatin in living cells: it displays a slow nuclear dynamics due to its anchoring to the DNA minor groove via the arginine-5 residue of the homeodomain. HOXC13 binds in vivo to the lamin B2 origin in a cell-cycle-dependent manner consistent with origin function; the interaction maps with nucleotide precision within the replicative complex. HOXC13 displays in vitro affinity for other replicative complex proteins; it interacts also in vivo with the same proteins in a cell-cycle-dependent fashion. Chromatin-structure modifying treatments, disturbing origin function, reduce also HOXC13-origin interaction. The described interactions are not restricted to a single origin nor to a single homeotic protein (also HOXC10 binds the lamin B2 origin in vivo). Thus, HOX complexes probably contribute in a general, structure-dependent manner, to origin identification and assembly of replicative complexes thereon, in presence of specific chromatin configurations.


Subject(s)
Homeodomain Proteins/physiology , Replication Origin , Animals , Cell Line , Chromatin/chemistry , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , HeLa Cells , Homeodomain Proteins/analysis , Homeodomain Proteins/metabolism , Humans , Lamin Type B/analysis , Mice , NIH 3T3 Cells
9.
Mol Cell Biol ; 30(6): 1382-96, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20065033

ABSTRACT

Cellular and biochemical studies support a role for all five human RecQ helicases in DNA replication; however, their specific functions during this process are unclear. Here we investigate the in vivo association of the five human RecQ helicases with three well-characterized human replication origins. We show that only RECQ1 (also called RECQL or RECQL1) and RECQ4 (also called RECQL4) associate with replication origins in a cell cycle-regulated fashion in unperturbed cells. RECQ4 is recruited to origins at late G(1), after ORC and MCM complex assembly, while RECQ1 and additional RECQ4 are loaded at origins at the onset of S phase, when licensed origins begin firing. Both proteins are lost from origins after DNA replication initiation, indicating either disassembly or tracking with the newly formed replisome. Nascent-origin DNA synthesis and the frequency of origin firing are reduced after RECQ1 depletion and, to a greater extent, after RECQ4 depletion. Depletion of RECQ1, though not that of RECQ4, also suppresses replication fork rates in otherwise unperturbed cells. These results indicate that RECQ1 and RECQ4 are integral components of the human replication complex and play distinct roles in DNA replication initiation and replication fork progression in vivo.


Subject(s)
DNA Replication , RecQ Helicases/metabolism , Cell Line , Cell Proliferation , Chromatin/metabolism , DNA/biosynthesis , DNA Replication Timing , Down-Regulation , G1 Phase , Humans , Lamin Type B/genetics , Lamin Type B/metabolism , Models, Biological , Protein Binding , RNA, Small Interfering/metabolism , Replication Origin/genetics , S Phase , beta-Globins/genetics , beta-Globins/metabolism
10.
Nat Struct Mol Biol ; 16(4): 412-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19343071

ABSTRACT

In eukaryotic cells, the cell-division cycle (CDC)-6 protein is essential to promote the assembly of pre-replicative complexes in the early G1 phase of the cell cycle, a process requiring tight regulation to ensure that proper origin licensing occurs once per cell cycle. Here we show that, in late G1 and early S phase, CDC6 is found in a complex also containing Cyclin A, cyclin-dependent kinase (CDK)-2 and the acetyltransferase general control nonderepressible 5 (GCN5). GCN5 specifically acetylates CDC6 at three lysine residues flanking its cyclin-docking motif, and this modification is crucial for the subsequent phosphorylation of the protein by Cyclin A-CDKs at a specific residue close to the acetylation site. GCN5-mediated acetylation and site-specific phosphorylation of CDC6 are both necessary for the relocalization of the protein to the cell cytoplasm in the S phase, as well as to regulate its stability. This two-step, intramolecular regulatory program by sequential modification of CDC6 seems to be essential for proper S-phase progression.


Subject(s)
Cell Cycle Proteins/metabolism , G1 Phase , Gene Expression Regulation , Nuclear Proteins/metabolism , S Phase , p300-CBP Transcription Factors/metabolism , Acetylation , Cell Line , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Humans , Lysine/metabolism , Models, Biological , Phosphorylation , Protein Binding
11.
Mol Cell ; 19(4): 567-75, 2005 Aug 19.
Article in English | MEDLINE | ID: mdl-16109380

ABSTRACT

The identification of metazoan origins of DNA replication has so far been hampered by the lack of a suitable genetic screening and by the cumbersomeness of the currently available mapping procedures. Here we describe the construction of a library of nascent DNA, representative of all cellular origin sequences, and its utilization as a screening probe for origin identification in large genomic regions. The procedure developed was successfully applied to the human 5q31.1 locus, encoding for the IL-3 and GM-CSF genes. Two novel origins were identified and subsequently characterized by competitive PCR mapping, located approximately 3.5 kb downstream of the GM-CSF gene. The two origins (GM-CSF Ori1 and Ori2) were shown to interact with different members of the DNA prereplication complex. This observation reinforces the universal paradigm that initiation of DNA replication takes place at, or in close proximity to, the binding sites of the trans-acting initiator proteins.


Subject(s)
Chromosome Mapping/methods , DNA Replication , DNA/genetics , Gene Library , Polymerase Chain Reaction/methods , Replication Origin , DNA/biosynthesis , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Evolution, Molecular , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-3/genetics , Nucleic Acid Hybridization
12.
Cell Cycle ; 4(7): 930-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15917652

ABSTRACT

Here we report a novel, noncompetitive mechanism that links acetylation and ubiquitination, in which the association of transcription factor E2F-1 with the cellular coactivator and acetyltransferase p300 determines its acetylation and subsequent ubiquitination. By using an antibody specifically recognizing the acetylated form of E2F-1 (AcE2F-1), we found that, after DNA damage, AcE2F-1 accumulates in the cells in a time-dependent manner, and that acetylation is increased by the expression of p300. Remarkably, the same DNA damaging conditions also induce the accumulation of ubiquitinated E2F-1, an event that is again markedly stimulated by p300 overexpression. The effects of p300 on E2F-1 ubiquitination require the integrity of the HAT domain of p300 and of the three acetylated lysines in E2F-1. Of note, p300-induced E2F-1 ubiquitination does not depend on the p45Skp2 E3 ligase, since it does not extend to other p45Skp2 targets and also occurs with an E2F-1 mutant devoid of the p45Skp2-binding domain but still retaining the acetylated region. Finally, p300-induced E2F-1 ubiquitination is not influenced by RB.


Subject(s)
DNA Damage , E2F1 Transcription Factor/metabolism , Ubiquitin/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Cell Cycle , Cell Line, Tumor , Cells, Cultured , DNA Damage/drug effects , Gene Expression Regulation , Humans , Lysine/metabolism , Protein Binding , Protein Processing, Post-Translational , SKP Cullin F-Box Protein Ligases/metabolism , Time Factors , p300-CBP Transcription Factors/chemistry
13.
J Cell Sci ; 117(Pt 22): 5221-31, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15454574

ABSTRACT

In eukaryotes, initiation of DNA replication requires the activity of the origin recognition complex (ORC). The largest subunit of this complex, Orc1p, has a critical role in this activity. Here we have studied the subnuclear distribution of the overexpressed human Orc1p during the cell cycle. Orc1p is progressively degraded during S-phase according to a spatio-temporal program and it never colocalizes with replication factories. Orc1p is resynthesized in G1. In early G1, the protein is distributed throughout the cell nucleus, but successively it preferentially associates with heterochromatin. This association requires a functional ATP binding site and a protein region partially overlapping the bromo-adjacent homology domain at the N-terminus of Orc1p. The same N-terminal region mediates the in vitro interaction with heterochromatin protein 1 (HP1). Fluorescence resonance energy transfer (FRET) experiments demonstrate the interaction of human Orc1p and HP1 in vivo. Our data suggest a role of HP1 in the recruitment but not in the stable association of Orc1p with heterochromatin. Indeed, the subnuclear distribution of Orc1p is not affected by treatments that trigger the dispersal of HP1.


Subject(s)
DNA-Binding Proteins , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Blotting, Western , COS Cells , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Chromatin/chemistry , DNA Replication , Fluorescence Resonance Energy Transfer , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Heterochromatin/chemistry , Heterochromatin/metabolism , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Models, Biological , Mutation , NIH 3T3 Cells , Origin Recognition Complex , Plasmids/metabolism , Polymerase Chain Reaction , Protein Binding , Protein Structure, Tertiary , Ribonuclease, Pancreatic/metabolism , S Phase , Time Factors , Transfection
14.
Cancer Gene Ther ; 9(1): 71-86, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11916246

ABSTRACT

In patients with chronic myelogenous leukemia (CML), abnormal expansion of myeloid cells is maintained by expression of the p210(bcr-abl) fusion protein. Thus, this protein and its mRNA represent primary targets to inhibit proliferation of these cells. Here we describe the properties of a ribozyme against the bcr-abl mRNA, expressed as a fusion transcript with the human U1 small nuclear RNA or the adenovirus VA1 RNA and delivered to the cells through retroviral vectors. These fusion ribozymes are specifically localized in the nucleus or in the cytoplasm, respectively. Transduction of 32D-LG7 myeloid cells, whose growth is IL-3 independent thanks to deregulated bcr-abl expression, imposed strong negative selective pressure on cell growth and induced restoration of an IL-3-dependent phenotype. Although expressed at a level similar to that of the U1-fusion ribozyme, the cytoplasmic VA1 ribozyme was a more powerful inhibitor of p210(bcr-abl) gene expression. In cells transduced with the vector expressing this ribozyme, the levels of the bcr-abl transcript were reduced up to 10(4)-fold, the p210(bcr-abl) protein became undetectable, and the cells underwent massive apoptosis when cultured in the absence of IL-3. Transduction of primary hematopoietic cells obtained from bone marrow of patients with CML resulted in remarkable reduction of bcr-abl mRNA levels, starting a few days after transduction. These results show the feasibility and efficacy of vector-expressed anti-bcr-abl ribozymes for purging of CML cells.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , RNA, Catalytic/therapeutic use , Animals , Apoptosis/physiology , Base Sequence , Blotting, Northern , Blotting, Western , Bone Marrow/metabolism , Cell Compartmentation/physiology , Flow Cytometry , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Genetic Therapy , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , Retroviridae/genetics , Transfection , Tumor Cells, Cultured/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...